You are on page 1of 11

See discussions, stats, and author profiles for this publication at: https://www.researchgate.

net/publication/259883815

Memory loss in Alzheimer's disease

Article · December 2013


Source: PubMed

CITATIONS READS

233 2,363

1 author:

Holger Jahn
AMEOS
234 PUBLICATIONS   6,642 CITATIONS   

SEE PROFILE

Some of the authors of this publication are also working on these related projects:

Circulating microRNAs in FTLD: Are there biomarkers? View project

Chloride channels View project

All content following this page was uploaded by Holger Jahn on 19 May 2015.

The user has requested enhancement of the downloaded file.


14 CH 8003 BA_INTERIEUR.qxd:DCNS#55 2/12/13 17:44 Page 445

Clinical research
Memory loss in Alzheimer’s disease
Holger Jahn, MD

Introduction

A sked to name a disease that affects “memory,”


most physicians would probably choose Alzheimer’s dis-
ease (AD). AD is the most common cause of dementia.
Currently, 30 million people worldwide suffer from
Alzheimer’s dementia and the World Health organiza-
tion projects that this number will triple over the next 20
Loss of memory is among the first symptoms reported by years.1 The cumulative incidence of Alzheimer dementia
patients suffering from Alzheimer’s disease (AD) and by has been estimated to rise from about 5% by age 70 to
their caretakers. Working memory and long-term declar- 50% by age 90, making it a very common disease.2
ative memory are affected early during the course of the Increasing longevity and demographic shifts in many
disease. The individual pattern of impaired memory societies will stress the health systems unless a cure for
functions correlates with parameters of structural or AD is found—or at least any therapy that postpones the
functional brain integrity. AD pathology interferes with onset of the dementia by 5 to 10 years for the time being.
the formation of memories from the molecular level to AD is a polygenetic neurodegenerative brain disorder
the framework of neural networks. The investigation of characterized by neocortical atrophy developing over
AD memory loss helps to identify the involved neural decades, showing the increasing loss of synapses and neu-
structures, such as the default mode network, the influ- rons first described by Alois Alzheimer in 1907.3
ence of epigenetic and genetic factors, such as ApoE4 Variations in the genes encoding the amyloid precursor
status, and evolutionary aspects of human cognition. proteins, presenilin-1 and presenilin-2, can directly cause
Clinically, the analysis of memory assists the definition of Alzheimer’s disease. The patients often display an early
AD subtypes, disease grading, and prognostic predic- onset of dementia in their forties, and these determinis-
tions. Despite new AD criteria that allow the earlier diag- tic genes affect whole families and patients with trisomy
nosis of the disease by inclusion of biomarkers derived 21. They, however, account for only 1% of all cases of
from cerebrospinal fluid or hippocampal volume analy- AD. The remaining 99% of sporadic AD occurs pre-
sis, neuropsychological testing remains at the core of AD
diagnosis. Author affiliations: University Hospital Hamburg-Eppendorf, Dept of
Psychiatry and Psychotherapy, Hamburg, Germany
© 2013, AICH – Servier Research Group Dialogues Clin Neurosci. 2013;15:445-454.

Address for correspondence: Holger Jahn, MD, University Hospital Hamburg-


Keywords: memory; Alzheimer’s disease; neuropsychological test; brain atrophy; Eppendorf, Dept of Psychiatry and Psychotherapy, Martinistr. 52, 20246
default mode network Hamburg, Germany
(e-mail: jahn@uke.de)

Copyright © 2013 AICH – Servier Research Group. All rights reserved 445 www.dialogues-cns.org
14 CH 8003 BA_INTERIEUR.qxd:DCNS#55 2/12/13 17:44 Page 446

Clinical research
Selected abbreviations and acronyms and finally frontal association cortices.12-14 The first lesions
DMN default mode network characteristic of AD appear in poorly myelinated limbic
DTI diffusion tensor imaging neurons in system areas related to memory and learning,
FA fractional anisotropy such as the hippocampus and the association cortex.
FX fragile X Highly myelinated neurons are only affected in the final
HARDI high angular resolution diffusion imaging phases of the disease.15 Low myelinization increases the
ICA independent components analysis overall energy expenditure of neurons. In addition, sub-
TBM tensor-based morphometry cortical neuron loss occurs in the nucleus basalis of
VBM voxel-based morphometry Meynert and the locus ceruleus, impairing the cholinergic
and noradrenergic transmitter systems in the neocor-
dominantly in patients older than 65 years.4 These tex.16,17 The parietal lobe, along with certain areas of the
patients also inherit 60% to 80% of their risk for late- prefrontal lobe, is one of the last areas of the human brain
onset AD, although it is not connected to the aforemen- to myelinate, and many of its neurons remain poorly
tioned genes, with the remainder being environmental.4,5 myelinated for the entire lifespan, which may explain their
In late-onset AD many different genes—some still vulnerability to factors capable of triggering AD.18-20 The
unidentified—are involved, each only attributing a minor atrophy runs slowly, but while in healthy aging only 0.2%
fraction of the individual’s overall risk, making it a pos- to 0.41% of the brain volume vanishes per year, the rates
sible example of antagonistic pleiotropy.6 Recently, sev- in AD may be ten times that, and in especially vulnerable
eral genome-wide association studies have identified new regions like the hippocampal formation atrophy rates
candidates (for reviews and databases see http://www.alz- might be even more devastatingly high and surpass 10%
gene.org). The best characterized genetic risk factor in per year (see also Figure 1).21-23 In terms of neuropsycho-
late-onset AD is the apolipoprotein E4 (ApoE4) geno-
type with ApoE4 carriers having a 4- to 10-fold increased
odds ratio of developing AD.7 The ApoE4 allele is an
ancestral variant. The human ApoE2 and ApoE3 alleles
are fairly recent mutations—less than 200 000 years old.8
ApoE3 is the most frequent allele, with frequencies of
about 60% in human populations. In contrast, ApoE4 has
a frequency of about 10%. This implies that ApoE3 and
ApoE2 must possess an advantage in order to have
gained prevalence in human populations so quickly; clin-
ical and preclinical data point to higher synaptic plastic-
ity and repair capacities in carriers of ApoE2 and ApoE3,
C Hippocampus left
D Lateral ventricle left
Lateral ventrical volume (mL)

which may explain the selection bias.9 The consumption 60


Hippocampal volume (mL)

and adaption of humans to a diet rich in meat and animal 2.5


50
fats can also be a reason for the selection of ApoE3.10
2
40
Atrophy in AD 1.5
30

1
The neuropathological hallmarks of the atrophy process 68 70 72 68 70 72
in AD are the presence of senile plaques (amyloid Age (y) Age (y)

deposits) and neurofibrillary tangles in autopsied brains.11


Figure 1. Atrophy in a case of AD over 4 years. (A) Reduction of gray mat-
Neurofibrillary tangles are composed of hyperphospho- ter (lateral view; corrected for age; P<0.05), (B) coronal view of
rylated tau protein located within neurons, whereas senile the left hippocampus at baseline and after 4 years (hippocam-
plaques are made up largely of amyloid-β species aggre- pal gray matter volume at T0 5.3±0.4 mL, at T4 3.5±0.2 mL),
gating in the extracellular space. These neuropathological (C) average hippocampal volume reduction of 0.2 mL per year
(–12%/year), (D) average increase of lateral ventricle volume of
changes start in the entorhinal cortex and hippocampal 2.7 mL/per year (+ 6.7%/year).
formations, later spreading into other temporal, parietal, (Courtesy of L. Spies, Jung-diagnostics GmbH, Hamburg, Germany)

446
14 CH 8003 BA_INTERIEUR.qxd:DCNS#55 2/12/13 17:44 Page 447

Memory loss in AD - Jahn Dialogues in Clinical Neuroscience - Vol 15 . No. 4 . 2013

logical tests, regional atrophy, and glucose metabolism time daily activities can no longer be performed properly.
correlate well with test results.24,25 Left hippocampal gray When brain atrophy progresses other psychiatric and
matter volume, for example, significantly correlates with neurological symptoms arise, and typically AD patients
performance in memory tasks, and left temporal gray lose weight and frequently develop difficulties in swal-
matter volume is related to performance in language lowing. This may lead to aspiration and subsequently
tasks. The rate of change in the left hippocampus corre- pneumonia, which is often the final cause of death in
lates with decline of performance in the Boston Naming demented patients.
Test Mini-Mental Status Examination, and the trailmak-
ing test B.24 Such analyses help with the definition of spe- The neuropsychology of AD:
cial AD subtypes like posterior cortical atrophy, or the tests and what they indicate
logopenic variant of AD.24-31 On the molecular level we
find a downregulation of synaptic genes across multiple Consensus exists that AD starts clinically with memory
brain regions and widespread proteomic signs of synaptic complaints, which may affect episodic memory, speech
stress or decay in the cerebrospinal fluid (CSF) or blood.32- production, with naming or semantic problems, or visual
34
Changes in the molecular fine structure of AD brains orientation. Memory can be defined as a process of
also arise independently of atrophy as resonance spec- encoding, storing, and retrieving information about outer
troscopic investigations in AD imply.35 and inner stimuli, or presentation of information to the
This review aims to highlight some aspects concerning nervous system of an organism that can be used to react
the development of memory deficits in AD that recently and position the organism towards new stimuli. Different
have or should have gained attention. categories of memory have been defined which also have
different neuroanatomical and neurophysiological cor-
Impact of new diagnostic criteria relates: short-term memory vs long-term memory or
implicit versus declarative memory. Short-term memory
Recently workgroups of the Alzheimer’s Association and is limited to just a few “chunks” in capacity, and lasts only
the National Institute on Aging have issued new criteria seconds to minutes.41 It depends on regions of the frontal
and guidelines to diagnose Alzheimer’s disease supplant- lobe and the parietal lobe. In contrast, long-term mem-
ing the previous guidelines first published in 1984.36-40 This ory seems almost limitless regarding its storage capaci-
marks a complete overhaul, and attemps to implement ties, for a potentially unlimited duration. It depends on
advances in our understanding of the disease in the way de novo protein synthesis and changes in the molecular
we diagnose the disease. The most notable differences are components of the neuronal networks involved in the
the use of biomarkers such as hippocampal atrophy, and specific cortical areas that can be attributed to different
the formalization of earlier disease stages before demen- memory types. Declarative memory, for example, can be
tia is apparent, such as mild cognitive impairment due to further subdivided into semantic memory, where context-
AD and the newly defined preclinical AD stage.38,39 While independent information is stored, and episodic memory,
the recommendations of the preclinical AD workgroup which stores information specific to a particular context,
are intended purely for research purposes and the aim of mainly time and place. Semantic memory is at first
diagnosing the disease earlier appears sensible since it is impaired in the language of AD patients, affecting verbal
likely that any intervention has to be started early to be fluency and naming. Semantic loss in AD may occur sev-
successful, it is also clear that we would almost all be eral years prior to diagnosis.42 The hippocampus is essen-
defined as having the disease using this definition, given tial to the consolidation of information from short-term
the increasing prevalence of AD in the very old. From a to long-term memory. Destruction of the hippocampal
scientific point of view, it might be more interesting to formation makes the storage of new memories impossi-
know why a few of us might not develop AD, even when ble. In the clinical context we use neuropsychological test
we are not dying from other diseases. As clinicians AD batteries like the CERAD (Consortium to Establish a
patients may first approach us with mere subjective con- Registry for Alzheimer’s Disease) examination, the Mini-
cerns about cognitive decline. This can develop into mild Mental State examination, and various other test con-
cognitive impairment with pathological neuropsycho- structs and scales, like the clinical dementia rating scale,
logical test results and progress into dementia, at which that investigate different aspects of memory over a broad

447
14 CH 8003 BA_INTERIEUR.qxd:DCNS#55 2/12/13 17:44 Page 448

Clinical research
range of various cognitive domains.43-45 Patients get pro- Regarding animal models, there are plenty of paradig-
filed in relation to tests they show abnormalities on, com- mata available to test memory functions, but there is an
pared with a healthy reference group adjusted for age overall lack of validated animal data that can be aligned
and education. AD patients typically display a cognitive with similar tests in human settings. Snigdha et al started
profile with impairments in multiple cognitive domains. with the comprehensive toolbox for Neurologic
This cognitive profile develops over time, and AD Behavioral Function from the National Institutes of
patients often start to show a progressive decay of work- Health (NIH) which contains evaluated tests for cogni-
ing memory. The patients display increased sensitivity to tive, motor, sensory, and emotional function for use in
distraction in memory tasks, the capacity of working epidemiologic and clinical studies spanning 3 to 85 years
memory measured, eg, digit span is, however, at first still of age and analyzed strengths and limitations of avail-
intact. Interestingly, the medications used currently to able animal behavioral tests to find matches. They
treat AD like acetylcholinesterase inhibitors or meman- defined a preclinical battery that aims to parallel the
tine work partly by increasing attention and concentra- NIH Toolbox, and may help to close the gap between
tion and work mainly in mild-to-moderate AD.46,47 data from different species.49
The deficits in attention and working memory associated
with damage to frontal subcortical circuits also influence Subjective cognitive impairment
executive functions in AD, impairing planning, problem
solving, and goal-directed behavior such as the ability to Subjective cognitive impairment without detectable
deploy response alternatives or modify behavior. AD objective memory deficit may no longer merely
patients show impaired results in tests that require plan- regarded as “normal aging” since it has been shown that
ning, problem solving, or cognitive flexibility, eg, the it is a major risk factor for the development of demen-
Wisconsin Card Sorting Test, the Stroop test, or the tia.50 A clear definition of what subjective memory
Tower of London Test. The manifestation of impairment impairment or subjective cognitive impairment actually
in such tests of executive functioning corresponds to the mean is currently lacking. An international task force is,
onset of difficulties in the performance of daily activities however, working on standard operating procedures that
in these patients and marks the progression to the state would enable comparable study designs. A consensus
of full dementia. The Boston Naming test assesses the regarding naming the concept “subjective cognitive
ability to name pictures of objects through spontaneous impairment” in view of previously used terminology
responses, and the need for various types of cueing. such as “subjective memory impairment” seems to be
Cued recall deficits are most closely associated with CSF arising. Subjective cognitive impairment is defined as the
biomarkers indicative of AD in subjects with mild cog- individual coming up with the mere feeling that some-
nitive impairment. This novel finding complements thing is not in order, without any objective parameters
results from prospective clinical studies and provides supporting that notion in the first place. Such a stage
further empirical support for cued recall as a specific labeled subjective cognitive impairment may precede
indicator of prodromal AD, in line with recently pro- mild cognitive impairment in the continuum of
posed research criteria.48 Another surprisingly simple Alzheimer disease manifestation. Using such a defini-
test is the Clock Drawing test. AD patients show early tion and without objective neuropsychological test alter-
difficulties in visuospatial processing and conceptual ations, the atrophy pattern of patients with subjective
errors like misrepresentation of numbers in the com- cognitive impairment seem to be related to the atrophy
mand, but not in the copy, condition, pointing to deficits pattern seen in AD.51 Individuals with subjective mem-
in semantic memory. The Trail Making Test A+B is a ory impairment showed greater similarity to an AD gray
neuropsychological test of visual attention measuring matter pattern, and episodic memory decline was asso-
mental processing speed, and the ability to switch ciated with an AD gray matter pattern in probands with
between different tasks. It consists of two parts in which subjective memory impairment.52 Patients with subjec-
the subject is asked to connect a set of 25 dots as fast as tive memory impairment also already showed hypome-
possible while maintaining accuracy. Visual search speed, tabolism in the right precuneus and hypermetabolism in
scanning and processing abilities, mental flexibility, and the right medial temporal lobe using (fludeoxyglucose
executive functioning can be assessed with this test. positron emission tomography, FDG-PET). Their gray

448
14 CH 8003 BA_INTERIEUR.qxd:DCNS#55 2/12/13 17:44 Page 449

Memory loss in AD - Jahn Dialogues in Clinical Neuroscience - Vol 15 . No. 4 . 2013

matter volume was reduced in the right hippocampus. At for decades and relates well to the finding that, for
follow-up, these patients showed poorer performance on example, plaque formation in AD starts in the entorhi-
measures of episodic memory. The observed memory nal cortex, the region also responsible for processing of
decline was associated with reduced glucose metabolism information on smell. A recent meta-analysis of 81 stud-
in the right precuneus at baseline. The authors conclude ies indicated that AD and PD patients are more
that their concept of subjective memory impairment impaired on odor identification and recognition tasks
may define the earliest clinical manifestation of AD.53 In than on odor detection threshold tasks. AD patients
another study patients with subjective memory under- were found to be more impaired on higher-order olfac-
went an associative episodic memory task matching tory tasks involving specific cognitive processes.57 Odor
faces to professions, including encoding, recall, and identification and recognition tests can be easily imple-
recognition, and a working memory task during func- mented in cognitive test batteries to detect already sub-
tional magnetic resonance imaging (fMRI). They clinical cases of AD. The impairment of smell recogni-
showed a reduction in right hippocampal activation dur- tion is of clinical importance, as patients often report
ing episodic memory recall, still in the absence of per- malodorous sensations and changes in, eg, the taste of
formance deficits. This was accompanied by increased foods leading to behavioral alterations. Consequences
activation of the right dorsolateral prefrontal cortex. No may range from increasing malnutriton to the develop-
such differences in performance and brain activation ment of delusions of poisoning that may trigger aggres-
were detected for working memory. This may indicate sive behavior. The deterioration of the neural network
subtle early neuronal dysfunction on the hippocampal in the entorhinal cortex leads to an impairment in the
level and compensatory mechanisms that preserve mem- ability to store and retrieve different representations of
ory performance.54 smell, with the decaying network yielding increasingly
Regarding ApoE4, cognitively unimpaired young elderly “default values” that have the tendency to be of rather
with and without subjective memory impairment were unpleasant character. This feature of the neural network
tested on episodic memory and on tasks of speed and of smell memory reflects the evolutionary pressure
executive function. Medial temporal lobe volumetric towards the secure recognition of “bad” smells pointing
measures were calculated from MRI images. In the sub- to poisonous or rotten food that is pivotal for the sur-
jective memory impairment group, ApoE4 carriers per- vival of the organism.
formed worse on the episodic memory and showed
smaller left hippocampal volumes. In the individuals AD and epileptic activity
without memory complaints, the ApoE4 carriers per-
formed better on episodic memory and had larger right The incidence of unprovoked seizures is clearly higher
hippocampal volumes (P=0.039). The interaction of in sporadic AD than in reference populations with impli-
group and ApoE genotype was significant for episodic cations for memory functions. Nonconvulsive epilepti-
memory and right and left hippocampal volumes. The form activity could underlie at least some of the cogni-
negative effect of ApoE4 on episodic memory and hip- tive impairments observed in AD. Up to 1 in 5 patients
pocampal volume in the group suffering from subjective with sporadic AD has at least 1 unprovoked clinically
memory decline also supports the notion that this may apparent seizure during their illness, and clinical guide-
be a prodromal condition of AD.55 In conclusion, the lines recommend obligatory treatment for this condition.
mere subjective feeling of being cognitively altered com- The risk of epileptic activity is greater in early-onset AD.
pared with the individual’s reference past can already be Many mutations in the presenilin-1 gene are associated
accompanied by subtle brain changes that if ongoing with epilepsy. Trisomy-21 patients with early-onset AD
may herald increasing memory decline in the future. also have frank seizures in 84% of cases. Many patients
with AD show fluctuations in cognitive functions such as
Memory of smell transient episodes of amnestic wandering or disorienta-
tion. While an intermittent inability to retrieve memo-
Impaired sense of smell or hyposmia is one of the ear- ries cannot be easily explained by relatively protracted
liest clinical features in neurodegenerative disorders like processes such as neuronal loss, plaque deposition, or
both AD or Parkinson’s disease.56 This has been known tangle formation, an abnormal epileptic activity of neu-

449
14 CH 8003 BA_INTERIEUR.qxd:DCNS#55 2/12/13 17:44 Page 450

Clinical research
ronal networks can. Extensive work in this field was impaired DMN consistent with metabolic and structural
published by the group of Lennart Mucke. They see the changes.67-69 The DMN is coupled with hippocampus dur-
possibility that high levels of β-amyloid induce epilepti- ing memory retrieval but not during memory encoding,
form activity, which triggers compensatory inhibitory pointing to the special positioning of the hippocampus
responses to counteract overexcitation that lead to between short-term and long-term memory.70 Encoding
changes in synaptic circuitry and an increase in structures of the DMN are among the first to show accu-
inhibitory activity in, eg, the temporal cortex. This leads mulation of β-amyloid even before symptoms emerge and
to changes in the texture of the neural networks images of β-amyloid plaques taken at the earliest stages of
involved and might explain disruptions of the networks AD show a distribution that is remarkably similar to the
as seen in the default mode network (DMN) in AD.58-60 anatomy of the default network.71 Buckner et al speculate
Transynaptic progression of toxicity effects of β-amyloid that AD pathology forms preferentially throughout the
inducing epileptic activity from the entorhinal cortex to DMN and may be linked to DMN activity.63 Their basic
other brain regions may explain cognitive dysfunctions idea is that the DMN’s continuous activity augments an
in AD.61 activity-dependent or metabolism-dependent cascade that
starts the β-amyloid cascade in these brain regions. Hence,
The default mode network memory would be affected preferentially by the disease
because the DMN is mainly relying on cortical structures
AD affects the default mode network (DMN). This net- that are also vital to memory functions and “burns” them
work comprises brain regions that are active and inter- during activity. Interestingly, ApoE4 carriers have also
connected in a wakeful state when the mind is not been found to have a higher rate of activity in the DMN
focused on something specific. Anatomically it includes at rest compared with ApoE2 or ApoE3 carriers, and
part of the medial temporal lobe, the medial prefrontal decreased connectivity.72-74 A successful connection of this
cortex, the posterior cingulate cortex, ventral precuneus, hypothesis with the β-amyloid hypothesis of AD may
and the medial, lateral, and inferior parietal cortex. This require any kind of upregulation of β-amyloid during
networks develops during childhood and adolescence neural activity. Indeed, there are some studies that may
and reaches full integration in adults, characterized by support such a link. Cirrito et al showed that β-amyloid
coherent infraslow EEG oscillations smaller than 0.1 Hz. increased following stimulation of the brain in mice
The DMN is linked to other low-frequency resting state expressing human amyloid precursor protein. They
networks in the brain and is anti-correlated with the ven- demonstrated that β-amyloid in the brain interstitial fluid
tral and dorsal attention network. Measurements of glu- was dynamically and directly influenced by synaptic activ-
cose metabolism with positron emission tomography ity on a timescale of minutes to hours.75 This observation
(PET), of structural atrophy with MRI, and intrinsic and suggests that synaptic activity can increase the presence of
task-evoked brain activity with fMRI in AD all suggest extracellular β-amyloid. A further supporting observations
an increasing disruption in the DMN.62 come from a new PET method of mapping glycolysis
When AD patients undergo a FDG-PET the pattern of based on measuring the ratio of oxygen to glucose con-
hypometabolism often mirrors the same regions that sumption. Vlassenko et al calculated the spatial distribu-
belong to the posterior parts of the DMN, namely the pos- tion of the regional glucose use apart from that entering
terior cingulate cortex, the retrosplenial cortex, inferior oxidative phosphorylation.76 The so-called “aerobic gly-
parietal lobule, and the lateral temporal cortex.63 Such colysis,” the process by which glucose is metabolized into
hypometabolism correlates with the mental status while cellular energy, might be more closely associated with neu-
AD progresses.64 Probands with a genetic risk for AD of ronal or synaptic activity than the mere glucose utilization.
being homozygous for ApoE4 develop this hypometabo- In humans, aerobic glycolysis represents 35% of the glu-
lism already quite early in the course of the disease.62,65 cose turnover in the brain of a newborn and 19% of the
Disruption in the DMN at the preclinical stages of the dis- glucose used in the brain of an alert adult.77 Certain asso-
ease by accelerated cortical atrophy affects the medial ciation areas in the human brain retain elevated levels of
temporal lobe and the posterior cingulum and the retros- aerobic glycolysis in adulthood related to cognitive func-
plenial cortex.63,66 Also, analysis of task-induced deactiva- tions such as the dorsolateral prefrontal cortex, which is
tion and analysis of intrinsic activity correlations show an associated with working memory, and the ventromedial

450
14 CH 8003 BA_INTERIEUR.qxd:DCNS#55 2/12/13 17:44 Page 451

Memory loss in AD - Jahn Dialogues in Clinical Neuroscience - Vol 15 . No. 4 . 2013

prefrontal cortex, the dorsomedial prefrontal cortex, the negative correlations were detected principally in para-
posterior cingulate cortex, the inferior parietal lobe, the median heteromodal cortices whereas positive correla-
lateral temporal cortex, and the hippocampus.62,76-79 In nor- tions were seen in cerebellum, thalamus, basal ganglia,
mal young adults aerobic glycolysis correlated positively lateral neocortices, and hippocampus.84 Sleep has adap-
and spatially with β-amyloid deposition observed in indi- tive and recreating functions that uphold waking activity
viduals with Alzheimer’s dementia and cognitively normal in humans and mammals in general. Our understanding
participants with already elevated β-amyloid levels, sug- of DMN activity and its regulation during sleep may be
gesting a possible link between regional aerobic glycolysis also important for our general understanding of phe-
in young adulthood and later development of AD pathol- nomena like memory, arousal, and consciousness.80,84-86
ogy. The map of resting-state glycolysis correlated remark- Clinically sleep-wake disturbances such as increased
ably well with the distribution of amyloid plaques.76 On the inadvertent daytime napping and insomnia at night affect
other side DMN activity and coherence is diminished dur- 25% to 40% of patients with mild-to-moderate AD.87
ing deep sleep. Here only partial network involvement was Even in mild cognitive impairment there are already
observed, with apparent decoupling of frontal areas from abnormalities in sleep architecture and electroen-
the DMN.80 An important study by Kang et al used in vivo cephalography measures. Sleep changes in patients with
micro-dialysis in mice and found that the amount of β- amnestic mild cognitive impairment may contribute to
amyloid in the interstitial brain fluid correlated positively memory deficits by interfering with sleep-dependent
with wakefulness. The amount of interstitial β-amyloid also memory consolidation.88
significantly increased during acute sleep deprivation. In a small study, Ju investigated sleep in 145 cognitively
Furthermore, chronic sleep restriction significantly healthy probands older than 45 years. Amyloid deposi-
increased, and a dual orexin receptor antagonist decreased tion, as assessed by β-amyloid levels, was present in 32
β-amyloid plaque formation in human amyloid precursor participants. This group had worse sleep quality, as mea-
protein transgenic mice.81 Interestingly, sleep deprivation, sured by sleep efficiency compared with those without
which is known to impair memory storage and retrieval, amyloid deposition, after correction for age, sex, and
reduces default mode network connectivity and anti-cor- ApoE4 allele carrier status, while the quantity of sleep
relation with the default attention networks during rest did not differ between groups. Frequent napping, 3 or
and task performance.82 Regarding AD subtypes, more days per week, was associated with amyloid depo-
Lehmann et al report that the posterior DMN and pre- sition. The authors concluded that indices for amyloid
cuneus network are commonly affected in all AD variants, deposition in the preclinical stage of AD appears to be
whereas syndrome-specific neurodegenerative patterns associated with worse sleep quality.89
are driven by the involvement of specific networks outside Taken together the brain activity patterns may directly
the DMN and characterize differentially early-onset AD modulate the molecular cascades that are relevant to dis-
(anterior salience network), logopenic variant of AD (lan- eases. In the case of AD, increased resting-state activity
guage network), and posterior cortical atrophy (visual net- may accelerate the formation of amyloid pathology.
work).83 This opens up perspectives for new interventions that
may take the form of a therapy that attempts to modify
Sleep and memory glycolysis or other aspects of brain metabolism or to
boost prophylaxis by the promotion of healthy sleep
Sleep and the functional connectome are overlapping behavior or working behavior.
research areas. Neuroimaging studies of sleep based on
EEG-PET and EEG-fMRI are revealing the brain net- Conclusion
works that support sleep. Such infraslow oscillations may
organize sleep-dependent neuroplastic processes includ- For the clinician the easily applicable neuropsycholog-
ing consolidation of episodic memory, for example. ical test batteries augmented by a test for olfactory
Picchioni et al found positive correlations between the recognition continue to be at the heart of the diagnos-
power in the infraslow EEG band and MRI blood oxy- tic process for dementias, although new methods like
gen level-dependent (BOLD) response in subcortical CSF analyses or MRI volumetry are increasingly avail-
regions and negative correlations in the cortex. Robust able and validated for clinical use. The use of amyloid-

451
14 CH 8003 BA_INTERIEUR.qxd:DCNS#55 2/12/13 17:44 Page 452

Clinical research
tracer PET or fMRI to investigate, for example, the many years before any relevant impairment is experi-
DMN are still more preclinical than clinical character. enced by the patient. In light of the fact that most of us
Despite enormous progress in our knowledge of some would suffer from AD, were we only to get old enough,
pathophysiologic mechanisms in the last decade, we are one question arises: when the gods put the diagnosis
still far from understanding AD and also probably from before the therapy, how much “before” did they actu-
finding a cure. The available medications or medications ally mean? As a clinician, I would appreciate any mea-
may, however, help us to manage some symptoms for sure of “momentum” that would help me to translate
our patients, and gain some time for them. To date the biomarker findings, eg, a positive amyloid scan in PET,
recent scientific advancements primarily offer earlier to the patient. Perhaps CSF tau concentrations could be
diagnosis. With new diagnostic criteria properly applied, helpful, but at the moment only time will tell us the indi-
we will likely be able to diagnose Alzheimer’s disease vidual course of the disease. ❏

REFERENCES 19. Jacobs HI, Van Boxtel MP, Jolles J, Verhey FR, Uylings HB. Parietal cor-
tex matters in Alzheimer's disease:an overview of structural, functional and
1. Wimo A, Jonsson L, Winblad B. An estimate of the worldwide preva- metabolic findings. Neurosci Biobehav Rev. 2012;36:297-309.
lence and direct costs of dementia in 2003. Dement Geriatr Cogn Dis. 20. Jacobs HI, van Boxtel MP, Gronenschild EH, et al. Patterns of gray and
2006;21:175-181. white matter changes in individuals at risk for Alzheimer's disease. Curr
2. Hebert LE, Scherr PA, Beckett LA, et al. Age-specific incidence of Alzheimer Res. 2012;9:1097-1105.
Alzheimer's disease in a community population. JAMA. 1995;273:1354-1359. 21. Fox NC, Freeborough PA, Rossor MN. Visualisation and quantification
3. Alzheimer A. Über eine eigenartige Erkrankung der Hirnrinde. of rates of atrophy in Alzheimer's disease. Lancet. 1996;348:94-97.
Allgemeine Zeitschrift für Psychiatrie. 1907;64:146-148. 22. Fox NC, Cousens S, Scahill R, Harvey RJ, Rossor MN. Using serial regis-
4. Blennow K, de Leon MJ, Zetterberg H. Alzheimer's disease. Lancet. tered brain magnetic resonance imaging to measure disease progression in
2006;368:387-403. Alzheimer disease:power calculations and estimates of sample size to detect
5. Gerrish A, Russo G, Richards A, et al. The role of variation at A beta PP, treatment effects. Arch Neurol. 2000;57:339-344.
PSEN1, PSEN2, and MAPT in late onset Alzheimer's disease. J Alzheimers Dis. 23. Andrews KA, Modat M, Macdonald KE, et al. Atrophy rates in asymp-
2012;28:377-387. tomatic amyloidosis:implications for Alzheimer prevention trials. PLoS One.
6. Williams GC. Pleiotropy, natural selection, and the evolution of senes- 2013;8:e58816.
cence. Evolution. 1957;11:398-411. 24. Arlt S, Buchert R, Spies L, Eichenlaub M, Lehmbeck JT, Jahn H.
7. Mayeux R. Gene-environment interaction in late-onset Alzheimer dis- Association between fully automated MRI-based volumetry of different
ease:the role of apolipoprotein-epsilon4. Alzheimer Dis Assoc Disord. brain regions and neuropsychological test performance in patients with
1998;12(suppl 3):S10-S15. amnestic mild cognitive impairment and Alzheimer's disease. Eur Arch
8. Fullerton SM, Clark AG, Weiss KM, et al. Apolipoprotein E variation at Psychiatry Clin Neurosci. 2013;263:335-344.
the sequence haplotype level:implications for the origin and maintenance 25. Arlt S, Brassen S, Wilke F, et al. Association between FDG uptake, CSF
of a major human polymorphism. Am J Hum Genet. 2000;67:881-900. biomarkers and cognitive performance in patients with probable
9. Bufill E, Carbonell E. Apolipoprotein E polymorphism and neuronal Alzheimer's disease. Pharmacopsychiatry. 2009;42:210-210.
plasticity. Am J Hum Biol. 2006;18:556-558. 26. Crutch SJ, Lehmann M, Schott JM, Rabinovici GD, Rossor MN, Fox NC.
10. Finch CE, Stanford CB. Meat-adaptive genes and the evolution of Posterior cortical atrophy. Lancet Neurol. 2012;11:170-178.
slower aging in humans. Q Rev Biol. 2004;79:3-50. 27. Crutch SJ, Schott JM, Rabinovici GD, et al. Shining a light on posterior
11. Braak H, Braak E. Neurofibrillary changes confined to the entorhinal cortical atrophy. Alzheimers Dement. 2013;9:463-465.
region and an abundance of cortical amyloid in cases of presenile and senile 28. Crutch SJ, Lehmann M, Warren JD, Rohrer JD. The language profile of
dementia. Acta Neuropathologica. 1990;80:479-486. posterior cortical atrophy. J Neurol Neurosurg Psychiatry. 2013;84:460-466.
12. Braak H, Braak E. Development of Alzheimer-related neurofibrillary 29. Lehmann M, Crutch SJ, Ridgway GR, et al. Cortical thickness and voxel-
changes in the neocortex inversely recapitulates cortical myelogenesis. Acta based morphometry in posterior cortical atrophy and typical Alzheimer's
Neuropathologica. 1996;92:197-201. disease. Neurobiol Aging. 2011;32:1466-1476.
13. Braak H, Braak E. Neuropathological staging of Alzheimer-related 30. Gorno-Tempini ML, Dronkers NF, Rankin KP, et al. Cognition and
changes. Acta Neuropathologica. 1991;82:239-259. anatomy in three variants of primary progressive aphasia. Ann Neurol.
14. Hyman BT, Vanhoesen GW, Damasio AR, Barnes CL. Alzheimers-disease 2004;55:335-346.
- cell-specific pathology isolates the hippocampal formation. Science. 31. Gorno-Tempini ML, Hillis AE, Weintraub S, et al. Classification of pri-
1984;225:1168-1170. mary progressive aphasia and its variants. Neurology. 2011;76:1006-1014.
15. Braak H, Del Tredici K, Schultz C, Braak E. Vulnerability of select neu- 32. Berchtold NC, Coleman PD, Cribbs DH, Rogers J, Gillen DL, Cotman CW.
ronal types to Alzheimer's disease. Ann N Y Acad Sci. 2000;924:53-61. Synaptic genes are extensively downregulated across multiple brain regions
16. Bondareff W, Mountjoy CQ, Roth M. Loss of neurons of origin of the in normal human aging and Alzheimer's disease. Neurobiol Aging.
adrenergic projection to cerebral cortex (nucleus locus ceruleus) in senile 2013;34:1653-1661.
dementia. Neurology. 1982;32:164-168. 33. Jahn H, Wittke S, Zuerbig P, et al. Peptide fingerprinting of Alzheimer's
17. Mann DM, Yates PO, Marcyniuk B. A comparison of changes in the disease in cerebrospinal fluid: identification and prospective evaluation of
nucleus basalis and locus caeruleus in Alzheimer's disease. J Neurol Neurosurg new synaptic biomarkers. Plos One. 2011;6.
Psychiatry. 1984;47:201-203. 34. Zuerbig P, Jahn H. Use of proteomic methods in the analysis of human
18. Jacobs HI, Van Boxtel MP, Uylings HB, Gronenschild EH, Verhey FR, Jolles body fluids in Alzheimer research. Electrophoresis. 2012;33:3617-3630.
J. Atrophy of the parietal lobe in preclinical dementia. Brain Cogn. 35. Jessen F, Guer O, Block W, et al. A multicenter H-1-MRS study of the
2011;75:154-163. medial temporal lobe in AD and MCI. Neurology. 2009;72:1735-1740.

452
14 CH 8003 BA_INTERIEUR.qxd:DCNS#55 2/12/13 17:44 Page 453

Memory loss in AD - Jahn Dialogues in Clinical Neuroscience - Vol 15 . No. 4 . 2013

Pérdida de memoria en la Enfermedad de La perte de mémoire dans la maladie


Alzheimer d’Alzheimer

La pérdida de memoria está entre los primeros sín- La perte de mémoire est un des premiers symp-
tomas referidos por los pacientes que padecen tômes rapportés par les patients souffrant de mala-
Enfermedad de Alzheimer (EA) y por sus cuidado- die d’Alzheimer (MA) et leurs soignants. La
res. La memoria de trabajo y la memoria declara- mémoire de travail et la mémoire déclarative à
tiva de largo plazo se afectan precozmente long terme sont touchées de façon précoce au
durante el curso de la enfermedad. El perfil indivi- cours de l’évolution de la maladie. Le schéma indi-
dual de deterioro de las funciones de la memoria viduel de la détérioration des fonctions mnésiques
se correlaciona con parámetros de integridad cere- correspond aux paramètres de l’intégrité cérébrale
bral estructural o funcional. La patología de la EA fonctionnelle ou structurelle. La pathologie de la
interfiere con la formación de memorias desde el MA interfère avec la formation de souvenirs allant
nivel molecular hasta el sistema de redes neurales. du niveau moléculaire jusqu’au cadre des réseaux
La investigación de la pérdida de memoria en la EA neuronaux. La recherche de la perte de mémoire
ayuda a identificar las estructuras neurales involu- de la MA aide à identifier les structures neuronales
cradas, como la red neural por defecto, la influen- impliquées, comme le réseau du « mode par
cia de factores epigenéticos y genéticos, como el défaut », l’influence des facteurs épigénétiques et
estado de la ApoE4, y aspectos evolucionistas de la génétiques, comme l’état de l’ApoE4 et les aspects
cognición humana. Clínicamente, el análisis de la évolutifs de la cognition humaine. Cliniquement,
memoria ayuda a la definición de los subtipos de l’analyse de la mémoire facilite la définition des
EA, a la clasificación de la enfermedad y a las pre- sous-types de MA, le stade de la maladie et la pré-
dicciones pronósticas. A pesar de los nuevos crite- vision du pronostic. Malgré les nouveaux critères
rios de EA que permiten un diagnóstico más pre- de MA permettant le diagnostic précoce de la
coz de la enfermedad mediante la incorporación maladie en faisant appel à des biomarqueurs déri-
de biomarcadores derivados del líquido céfalo vés du liquide céphalo-rachidien ou à l’analyse du
raquídeo o del análisis del volumen del hipocampo, volume de l’hippocampe, les tests neuropsycholo-
las pruebas neuropsicológicas persisten en lo giques restent au centre du diagnostic de la MA.
nuclear del diagnóstico de EA.

36. Jack CR, Jr, Albert MS, Knopman DS, et al. Introduction to the recom- 43. Morris JC. Clinical dementia rating:a reliable and valid diagnostic and
mendations from the National Institute on Aging-Alzheimer's Association staging measure for dementia of the Alzheimer type. Int Psychogeriatr.
workgroups on diagnostic guidelines for Alzheimer's disease. Alzheimers 1997;9(suppl 1):173-176;discussion 177-178.
Dementia. 2011;7:257-262. 44. Morris JC, Heyman A, Mohs RC, et al. The consortium to establish a reg-
37. McKhann GM, Knopman DS, Chertkow H, et al. The diagnosis of istry for Alzheimer's Disease (CERAD). Part I. Clinical and neuropsychologi-
dementia due to Alzheimer's disease: recommendations from the National cal assessment of Alzheimer's disease. Neurology. 1989;39:1159-1165.
Institute on Aging-Alzheimer's Association workgroups on diagnostic guide- 45. Morris JC, Mohs RC, Rogers H, Fillenbaum G, Heyman A. Consortium to
lines for Alzheimer's disease. Alzheimers Dementia. 2011;7:263-269. establish a registry for Alzheimer's disease (CERAD): clinical and neuropsy-
38. Sperling RA, Aisen PS, Beckett LA, et al. Toward defining the preclini- chological assessment of Alzheimer's disease. Psychopharmacol Bull.
cal stages of Alzheimer's disease: recommendations from the National 1988;24:641-652.
Institute on Aging-Alzheimer's Association workgroups on diagnostic guide- 46. Pepeu G, Giovannini MG. Cholinesterase inhibitors and memory. Chem
lines for Alzheimer's disease. Alzheimers Dementia. 2011;7:280-292. Biol Interact. 2010;187:403-408.
39. Albert MS, DeKosky ST, Dickson D, et al. The diagnosis of mild cogni- 47. Pepeu G, Giovannini MG, Bracco L. Effect of cholinesterase inhibitors
tive impairment due to Alzheimer's disease: recommendations from the on attention. Chem Biol Interact. 2013;203:361-364.
National Institute on Aging-Alzheimer's Association workgroups on diag- 48. Wagner M, Wolf S, Reischies FM, et al. Biomarker validation of a cued recall
nostic guidelines for Alzheimer's disease. Alzheimers Dementia. 2011;7:270- memory deficit in prodromal Alzheimer disease. Neurology. 2012;78:379-386.
279. 49. Snigdha S, Milgram NW, Willis SL, et al. A preclinical cognitive test bat-
40. Hyman BT, Phelps CH, Beach TG, et al. National Institute on Aging- tery to parallel the National Institute of Health Toolbox in humans:bridg-
Alzheimer's Association guidelines for the neuropathologic assessment of ing the translational gap. Neurobiol Aging. 2013;34:1891-1901.
Alzheimer's disease. Alzheimers Dementia. 2012;8:1-13. 50. Jessen F, Wiese B, Bachmann C, et al. Prediction of dementia by sub-
41. Miller GA. The magical number seven, plus or minus two: some limits jective memory impairment effects of severity and temporal association
on our capacity for processing information. Psychol Rev.1956;63:81-97. with cognitive impairment. Arch Gen Psychiatry. 2010;67:414-422.
42. Verma M, Howard RJ. Semantic memory and language dysfunction in 51. Jessen F, Feyen L, Freymann K, et al. Volume reduction of the entorhinal
early Alzheimer's disease: a review. Int J Geriatr Psychiatry. 2012;27:1209-1217. cortex in subjective memory impairment. Neurobiol Aging. 2006;27:1751-1756.

453
14 CH 8003 BA_INTERIEUR.qxd:DCNS#55 2/12/13 17:44 Page 454

Clinical research
52. Peter J, Scheef L, Abdulkadir A, et al. Gray matter atrophy pattern in 70. Huijbers W, Pennartz CM, Cabeza R, Daselaar SM. The hippocampus is
elderly with subjective memory impairment. Alzheimers Dement. 2013 Jul 15. coupled with the default network during memory retrieval but not during
[Epub ahead of print]. memory encoding. PLoS One. 2011;6:e17463.
53. Scheef L, Spottke A, Daerr M, et al. Glucose metabolism, gray matter 71. Klunk WE, Engler H, Nordberg A, et al. Imaging brain amyloid in
structure, and memory decline in subjective memory impairment. Neurology. Alzheimer's disease with Pittsburgh Compound-B. Ann Neurol. 2004;55:306-
2012;79:1332-1339. 319.
54. Erk S, Spottke A, Meisen A, Wagner M, Walter H, Jessen F. Evidence of 72. Sheline YI, Morris JC, Snyder AZ, et al. APOE4 allele disrupts resting
neuronal compensation during episodic memory in subjective memory state fMRI connectivity in the absence of amyloid plaques or decreased CSF
impairment. Arch Gen Psychiatry. 2011;68:845-852. Aβ42. J Neurosci. 2010;30:17035-17040.
55. Striepens N, Scheef L, Wind A, et al. Interaction effects of subjective 73. Patel KT, Stevens MC, Pearlson GD, et al. Default mode network activ-
memory impairment and ApoE4 genotype on episodic memory and hip- ity and white matter integrity in healthy middle-aged ApoE4 carriers. Brain
pocampal volume. Psychol Med. 2011;41:1997-2006. Imaging Behav. 2013;7:60-67.
56. Peters JM, Hummel T, Kratzsch T, Lötsch J, Skarke C, Frölich L. Olfactory 74. Filippini N, MacIntosh BJ, Hough MG, et al. Distinct patterns of brain
function in mild cognitive impairment and Alzheimer's disease:an investi- activity in young carriers of the APOE-epsilon4 allele. Proc Natl Acad Sci U S A.
gation using psychophysical and electrophysiological techniques. Am J 2009;106:7209-7214.
Psychiatry. 2003;160:1995-2002. 75. Cirrito JR, Yamada KA, Finn MB, et al. Synaptic activity regulates inter-
57. Rahayel S, Frasnelli J, Joubert S. The effect of Alzheimer's disease and stitial fluid amyloid-beta levels in vivo. Neuron. 2005;48:913-922.
Parkinson's disease on olfaction:a meta-analysis. Behav Brain Res. 76. Vlassenko AG, Vaishnavi SN, Couture L, et al. Spatial correlation
2012;231:60-74. between brain aerobic glycolysis and amyloid-β (Aβ) deposition. Proc Natl
58. Palop JJ, Mucke L. Epilepsy and cognitive impairments in Alzheimer dis- Acad Sci U S A. 2010;107:17763-17767.
ease. Arch Neurol. 2009;66:435-440. 77. Vaishnavi SN, Vlassenko AG, Rundle MM, Snyder AZ, Mintun MA,
59. Palop JJ, Mucke L. Synaptic depression and aberrant excitatory network Raichle ME. Regional aerobic glycolysis in the human brain. Proc Natl Acad
activity in Alzheimer's disease: two faces of the same coin? Neuromolecular Sci U S A. 2010;107:17757-17762.
Med. 2010;12:48-55.
78. Buckner RL. Human functional connectivity:new tools, unresolved ques-
60. Palop JJ, Mucke L, Roberson ED. Quantifying biomarkers of cognitive
tions. Proc Natl Acad Sci U S A. 2010;107:10769-10770.
dysfunction and neuronal network hyperexcitability in mouse models of
79. Buckner RL. The role of the hippocampus in prediction and imagina-
Alzheimer's disease: depletion of calcium-dependent proteins and inhibitory
tion. Annu Rev Psychol. 2010;61:27-48, C21-C28.
hippocampal remodeling. Methods Mol Biol. 2011;670:245-262.
80. Horovitz SG, Braun AR, Carr WS, et al. Decoupling of the brain's default
61. Harris JA, Devidze N, Verret L, et al. Transsynaptic progression of amy-
mode network during deep sleep. Proc Natl Acad Sci U S A. 2009;106:11376-
loid-β-induced neuronal dysfunction within the entorhinal-hippocampal
11381.
network. Neuron. 2010;68:428-441.
81. Kang JE, Lim MM, Bateman RJ, et al. Amyloid-beta dynamics are regu-
62. Buckner RL, Andrews-Hanna JR, Schacter DL. The brain's default net-
lated by orexin and the sleep-wake cycle. Science. 2009;326:1005-
work: anatomy, function, and relevance to disease. Ann N Y Acad Sci.
2008;1124:1-38. 1007.
63. Buckner RL, Snyder AZ, Shannon BJ, et al. Molecular, structural, and 82. De Havas JA, Parimal S, Soon CS, Chee MW. Sleep deprivation reduces
functional characterization of Alzheimer's disease: evidence for a relation- default mode network connectivity and anti-correlation during rest and
ship between default activity, amyloid, and memory. J Neurosci. task performance. Neuroimage. 2012;59:1745-1751.
2005;25:7709-7717. 83. Lehmann M, Madison CM, Ghosh PM, et al. Intrinsic connectivity net-
64. Herholz K, Salmon E, Perani D, et al. Discrimination between Alzheimer works in healthy subjects explain clinical variability in Alzheimer's disease.
dementia and controls by automated analysis of multicenter FDG PET. Proc Natl Acad Sci U S A. 2013;110:11606-11611.
Neuroimage. 2002;17:302-316. 84. Picchioni D, Horovitz SG, Fukunaga M, et al. Infraslow EEG oscillations
65. Reiman EM, Caselli RJ, Yun LS, et al. Preclinical evidence of Alzheimer's organize large-scale cortical-subcortical interactions during sleep:a com-
disease in persons homozygous for the epsilon 4 allele for apolipoprotein bined EEG/fMRI study. Brain Res. 2011;1374:63-72.
E. N Engl J Med. 1996;334:752-758. 85. Picchioni D, Duyn JH, Horovitz SG. Sleep and the functional connec-
66. Scahill RI, Schott JM, Stevens JM, Rossor MN, Fox NC. Mapping the evo- tome. Neuroimage. 2013;80:387-396.
lution of regional atrophy in Alzheimer's disease: unbiased analysis of fluid- 86. Chow HM, Horovitz SG, Carr WS, et al. Rhythmic alternating patterns
registered serial MRI. Proc Natl Acad Sci U S A. 2002;99:4703-4707. of brain activity distinguish rapid eye movement sleep from other states of
67. Lustig C, Snyder AZ, Bhakta M, et al. Functional deactivations: change consciousness. Proc Natl Acad Sci U S A. 2013;110:10300-10305.
with age and dementia of the Alzheimer type. Proc Natl Acad Sci U S A. 87. Moran M, Lynch CA, Walsh C, Coen R, Coakley D, Lawlor BA. Sleep dis-
2003;100:14504-14509. turbance in mild to moderate Alzheimer's disease. Sleep Med. 2005;6:347-
68. Celone KA, Calhoun VD, Dickerson BC, et al. Alterations in memory net- 352.
works in mild cognitive impairment and Alzheimer's disease:an indepen- 88. Westerberg CE, Mander BA, Florczak SM, et al. Concurrent impairments
dent component analysis. J Neurosci. 2006;26:10222-10231. in sleep and memory in amnestic mild cognitive impairment. J Int
69. Greicius MD, Srivastava G, Reiss AL, Menon V. Default-mode network Neuropsychol Soc. 2012;18:490-500.
activity distinguishes Alzheimer's disease from healthy aging:evidence from 89. Ju YE, McLeland JS, Toedebusch CD, et al. Sleep quality and preclinical
functional MRI. Proc Natl Acad Sci U S A. 2004;101:4637-4642. Alzheimer disease. JAMA Neurol. 2013;70:587-593.

454

View publication stats

You might also like